Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Perspective
  • Published:

OPINION

Molecular subtypes of small cell lung cancer: a synthesis of human and mouse model data

An Author Correction to this article was published on 07 June 2019

This article has been updated

Abstract

Small cell lung cancer (SCLC) is an exceptionally lethal malignancy for which more effective therapies are urgently needed. Several lines of evidence, from SCLC primary human tumours, patient-derived xenografts, cancer cell lines and genetically engineered mouse models, appear to be converging on a new model of SCLC subtypes defined by differential expression of four key transcription regulators: achaete-scute homologue 1 (ASCL1; also known as ASH1), neurogenic differentiation factor 1 (NeuroD1), yes-associated protein 1 (YAP1) and POU class 2 homeobox 3 (POU2F3). In this Perspectives article, we review and synthesize these recent lines of evidence and propose a working nomenclature for SCLC subtypes defined by relative expression of these four factors. Defining the unique therapeutic vulnerabilities of these subtypes of SCLC should help to focus and accelerate therapeutic research, leading to rationally targeted approaches that may ultimately improve clinical outcomes for patients with this disease.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Different nomenclature describing SCLC subtypes.
Fig. 2: Molecular subtypes of SCLC defined by expression of key transcription regulators.

Similar content being viewed by others

Change history

  • 07 June 2019

    An amendment to this paper has been published and can be accessed via a link at the top of the paper.

  • 27 June 2019

    An author correction to Supplementary Table 1 was made, but the original file was not updated. We have now updated Supplementary Table 1 in the original article.

References

  1. Gazdar, A. F., Bunn, P. A. & Minna, J. D. Small-cell lung cancer: what we know, what we need to know and the path forward. Nat. Rev. Cancer 17, 725–737 (2017).

    Article  CAS  Google Scholar 

  2. Sabari, J. K. et al. Unravelling the biology of SCLC: implications for therapy. Nat. Rev. Clin. Oncol. 14, 549–561 (2017).

    Article  CAS  Google Scholar 

  3. Hann, C. L. et al. in Cancer Principles & Practice of Oncology Ch. 49 (eds DeVita, V. T., Lawrence, T. S. & Rosenberg, S. A.) 671–700 (Wolters Kluwer, 2019).

  4. Horn, L. et al. First-line atezolizumab plus chemotherapy in extensive-stage small-cell lung cancer. N. Engl. J. Med. 379, 2220–2229 (2018).

    Article  CAS  Google Scholar 

  5. von Pawel, J. et al. Topotecan versus cyclophosphamide, doxorubicin, and vincristine for the treatment of recurrent small-cell lung cancer. J. Clin. Oncol. 17, 658–667 (1999).

    Article  Google Scholar 

  6. Ready, N. et al. Third-line nivolumab monotherapy in recurrent SCLC: CheckMate 032. J. Thorac. Oncol. 14, 237–244 (2018).

    Article  Google Scholar 

  7. Jordan, E. J. et al. Prospective comprehensive molecular characterization of lung adenocarcinomas for efficient patient matching to approved and emerging therapies. Cancer Discov. 7, 596–609 (2017).

    Article  CAS  Google Scholar 

  8. Gazdar, A. F. et al. Establishment of continuous, clonable cultures of small-cell carcinoma of lung which have amine precursor uptake and decarboxylation cell properties. Cancer Res. 40, 3502–3507 (1980).

    CAS  PubMed  Google Scholar 

  9. Carney, D. N. et al. Establishment and identification of small cell lung cancer cell lines having classic and variant features. Cancer Res. 45, 2913–2923 (1985).

    CAS  PubMed  Google Scholar 

  10. Gazdar, A. F. et al. Characterization of variant subclasses of cell lines derived from small cell lung cancer having distinctive biochemical, morphological, and growth properties. Cancer Res. 45, 2924–2930 (1985).

    CAS  PubMed  Google Scholar 

  11. Rudin, C. M. et al. Comprehensive genomic analysis identifies SOX2 as a frequently amplified gene in small-cell lung cancer. Nat. Genet. 44, 1111–1116 (2012).

    Article  CAS  Google Scholar 

  12. Peifer, M. et al. Integrative genome analyses identify key somatic driver mutations of small-cell lung cancer. Nat. Genet. 44, 1104–1110 (2012).

    Article  CAS  Google Scholar 

  13. George, J. et al. Comprehensive genomic profiles of small cell lung cancer. Nature 524, 47–53 (2015).

    Article  CAS  Google Scholar 

  14. Poirier, J. T. et al. Selective tropism of Seneca Valley virus for variant subtype small cell lung cancer. J. Natl Cancer Inst. 105, 1059–1065 (2013).

    Article  CAS  Google Scholar 

  15. Neptune, E. R. et al. Targeted disruption of NeuroD, a proneural basic helix-loop-helix factor, impairs distal lung formation and neuroendocrine morphology in the neonatal lung. J. Biol. Chem. 283, 21160–21169 (2008).

    Article  CAS  Google Scholar 

  16. Borges, M. et al. An achaete-scute homologue essential for neuroendocrine differentiation in the lung. Nature 386, 852–855 (1997).

    Article  CAS  Google Scholar 

  17. Ito, T. et al. Basic helix-loop-helix transcription factors regulate the neuroendocrine differentiation of fetal mouse pulmonary epithelium. Development 127, 3913–3921 (2000).

    CAS  PubMed  Google Scholar 

  18. Poirier, J. T. et al. DNA methylation in small cell lung cancer defines distinct disease subtypes and correlates with high expression of EZH2. Oncogene 34, 5869–5878 (2015).

    Article  CAS  Google Scholar 

  19. The Cancer Genome Atlas Research Network. Comprehensive genomic characterization of squamous cell lung cancers. Nature 489, 519–525 (2012).

    Article  Google Scholar 

  20. Christensen, C. L. et al. Targeting transcriptional addictions in small cell lung cancer with a covalent CDK7 inhibitor. Cancer Cell 26, 909–922 (2014).

    Article  CAS  Google Scholar 

  21. Lan, M. S. & Breslin, M. B. Structure, expression, and biological function of INSM1 transcription factor in neuroendocrine differentiation. FASEB J. 23, 2024–2033 (2009).

    Article  CAS  Google Scholar 

  22. Fujino, K. et al. Insulinoma-associated protein 1 is a crucial regulator of neuroendocrine differentiation in lung cancer. Am. J. Pathol. 185, 3164–3177 (2015).

    Article  CAS  Google Scholar 

  23. Gierl, M. S. et al. The zinc-finger factor Insm1 (IA-1) is essential for the development of pancreatic beta cells and intestinal endocrine cells. Genes Dev. 20, 2465–2478 (2006).

    Article  CAS  Google Scholar 

  24. Wildner, H. et al. Insm1 (IA-1) is a crucial component of the transcriptional network that controls differentiation of the sympatho-adrenal lineage. Development 135, 473–481 (2008).

    Article  CAS  Google Scholar 

  25. Borromeo, M. D. et al. ASCL1 and NEUROD1 reveal heterogeneity in pulmonary neuroendocrine tumors and regulate distinct genetic programs. Cell Rep. 16, 1259–1272 (2016).

    Article  CAS  Google Scholar 

  26. Gratton, M. O. et al. Hes6 promotes cortical neurogenesis and inhibits Hes1 transcription repression activity by multiple mechanisms. Mol. Cell. Biol. 23, 6922–6935 (2003).

    Article  CAS  Google Scholar 

  27. Wooten, D. et al. Small cell lung cancer subtypes identified by systems-level modeling of transcription factor networks. Preprint at bioRxiv https://www.biorxiv.org/content/10.1101/506402v2 (2018).

  28. Meuwissen, R. et al. Induction of small cell lung cancer by somatic inactivation of both Trp53 and Rb1 in a conditional mouse model. Cancer Cell 4, 181–189 (2003).

    Article  CAS  Google Scholar 

  29. Sutherland, K. D. et al. Cell of origin of small cell lung cancer: inactivation of Trp53 and Rb1 in distinct cell types of adult mouse lung. Cancer Cell 19, 754–764 (2011).

    Article  CAS  Google Scholar 

  30. Yang, D. et al. Intertumoral heterogeneity in SCLC is influenced by the cell type of origin. Cancer Discov. 8, 1316–1331 (2018).

    Article  CAS  Google Scholar 

  31. Schaffer, B. E. et al. Loss of p130 accelerates tumor development in a mouse model for human small-cell lung carcinoma. Cancer Res. 70, 3877–3883 (2010).

    Article  CAS  Google Scholar 

  32. Mollaoglu, G. et al. MYC drives progression of small cell lung cancer to a variant neuroendocrine subtype with vulnerability to aurora kinase inhibition. Cancer Cell 31, 270–285 (2017).

    Article  CAS  Google Scholar 

  33. Little, C. D. et al. Amplification and expression of the c-myc oncogene in human lung cancer cell lines. Nature 306, 194–196 (1983).

    Article  CAS  Google Scholar 

  34. McColl, K. et al. Reciprocal expression of INSM1 and YAP1 defines subgroups in small cell lung cancer. Oncotarget 8, 73745–73756 (2017).

    Article  Google Scholar 

  35. Huang, Y. H. et al. POU2F3 is a master regulator of a tuft cell-like variant of small cell lung cancer. Genes Dev. 32, 915–928 (2018).

    Article  CAS  Google Scholar 

  36. Yamashita, J. et al. Skn-1a/Pou2f3 functions as a master regulator to generate Trpm5-expressing chemosensory cells in mice. PLOS ONE 12, e0189340 (2017).

    Article  Google Scholar 

  37. Zhang, W. et al. Small cell lung cancer tumors and preclinical models display heterogeneity of neuroendocrine phenotypes. Transl Lung Cancer Res. 7, 32–49 (2018).

    Article  CAS  Google Scholar 

  38. The Cancer Genome Atlas Research Network. Comprehensive molecular profiling of lung adenocarcinoma. Nature 511, 543–550 (2014).

    Article  Google Scholar 

  39. Lim, J. S. et al. Intratumoural heterogeneity generated by Notch signalling promotes small-cell lung cancer. Nature 545, 360–364 (2017).

    Article  CAS  Google Scholar 

  40. Hodgkinson, C. L. et al. Tumorigenicity and genetic profiling of circulating tumor cells in small-cell lung cancer. Nat. Med. 20, 897–903 (2014).

    Article  CAS  Google Scholar 

  41. Drapkin, B. J. et al. Genomic and functional fidelity of small cell lung cancer patient-derived xenografts. Cancer Discov. 8, 600–615 (2018).

    Article  CAS  Google Scholar 

  42. Calbo, J. et al. A functional role for tumor cell heterogeneity in a mouse model of small cell lung cancer. Cancer Cell 19, 244–256 (2011).

    Article  CAS  Google Scholar 

  43. Kwon, M. C. et al. Paracrine signaling between tumor subclones of mouse SCLC: a critical role of ETS transcription factor Pea3 in facilitating metastasis. Genes Dev. 29, 1587–1592 (2015).

    Article  CAS  Google Scholar 

  44. Williamson, S. C. et al. Vasculogenic mimicry in small cell lung cancer. Nat. Commun. 7, 13322 (2016).

    Article  CAS  Google Scholar 

  45. Saunders, L. R. et al. A DLL3-targeted antibody-drug conjugate eradicates high-grade pulmonary neuroendocrine tumor-initiating cells in vivo. Sci. Transl Med. 7, 302ra136 (2015).

    Article  Google Scholar 

  46. Cardnell, R. J. et al. Protein expression of TTF1 and cMYC define distinct molecular subgroups of small cell lung cancer with unique vulnerabilities to aurora kinase inhibition, DLL3 targeting, and other targeted therapies. Oncotarget 8, 73419–73432 (2017).

    Article  Google Scholar 

  47. Jia, D. et al. Crebbp loss drives small cell lung cancer and increases sensitivity to HDAC inhibition. Cancer Discov. 8, 1422–1437 (2018).

    Article  Google Scholar 

  48. Takagi, S. et al. LSD1 inhibitor T-3775440 inhibits SCLC cell proliferation by disrupting LSD1 interactions with SNAG domain proteins INSM1 and GFI1B. Cancer Res. 77, 4652–4662 (2017).

    Article  CAS  Google Scholar 

  49. Augert, A. et al. Targeting NOTCH activation in small cell lung cancer through LSD1 inhibition. Sci. Signal. 12, eaau2922 (2019).

    Article  CAS  Google Scholar 

  50. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02034123 (2019).

  51. Rudin, C. M. et al. Phase I clinical study of Seneca Valley Virus (SVV-001), a replication-competent picornavirus, in advanced solid tumors with neuroendocrine features. Clin. Cancer Res. 17, 888–895 (2011).

    Article  CAS  Google Scholar 

  52. Belani, C. P. et al. Vismodegib or cixutumumab in combination with standard chemotherapy for patients with extensive-stage small cell lung cancer: a trial of the ECOG-ACRIN Cancer Research Group (E1508). Cancer 122, 2371–2378 (2016).

    Article  CAS  Google Scholar 

  53. Sen, T., Gay, C. M. & Byers, L. A. Targeting DNA damage repair in small cell lung cancer and the biomarker landscape. Transl Lung Cancer Res. 7, 50–68 (2018).

    Article  CAS  Google Scholar 

  54. Sen, T. et al. CHK1 inhibition in small-cell lung cancer produces single-agent activity in biomarker-defined disease subsets and combination activity with cisplatin or olaparib. Cancer Res. 77, 3870–3884 (2017).

    Article  CAS  Google Scholar 

  55. Udyavar, A. R. et al. Novel hybrid phenotype revealed in small cell lung cancer by a transcription factor network model that can explain tumor heterogeneity. Cancer Res. 77, 1063–1074 (2017).

    Article  CAS  Google Scholar 

  56. Hellmann, M. D. et al. Tumor mutational burden and efficacy of nivolumab monotherapy and in combination with ipilimumab in small-cell lung cancer. Cancer Cell 33, 853–861 (2018).

    Article  CAS  Google Scholar 

  57. Ott, P. A. et al. Pembrolizumab in patients with extensive-stage small-cell lung cancer: results from the phase Ib KEYNOTE-028 Study. J. Clin. Oncol. 35, 3823–3829 (2017).

    Article  CAS  Google Scholar 

  58. Sen, T. et al. Targeting DNA damage response promotes anti-tumor immunity through STING-mediated T cell activation in small cell lung cancer. Cancer Discov. https://doi.org/10.1158/2159-8290.CD-18-1020 (2019).

    Article  PubMed  Google Scholar 

  59. Skoulidis, F. et al. STK11/LKB1 mutations and PD-1 inhibitor resistance in KRAS-mutant lung adenocarcinoma. Cancer Discov. 8, 822–835 (2018).

    Article  CAS  Google Scholar 

  60. Sequist, L. V. et al. Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors. Sci. Transl Med. 3, 75ra26 (2011).

    Article  Google Scholar 

  61. Niederst, M. J. et al. RB loss in resistant EGFR mutant lung adenocarcinomas that transform to small-cell lung cancer. Nat. Commun. 6, 6377 (2015).

    Article  CAS  Google Scholar 

  62. Mu, P. et al. SOX2 promotes lineage plasticity and antiandrogen resistance in TP53- and RB1-deficient prostate cancer. Science 355, 84–88 (2017).

    Article  CAS  Google Scholar 

  63. Ku, S. Y. et al. Rb1 and Trp53 cooperate to suppress prostate cancer lineage plasticity, metastasis, and antiandrogen resistance. Science 355, 78–83 (2017).

    Article  CAS  Google Scholar 

  64. Rekhtman, N. et al. Next-generation sequencing of pulmonary large cell neuroendocrine carcinoma reveals small cell carcinoma-like and non-small cell carcinoma-like subsets. Clin. Cancer Res. 22, 3618–3629 (2016).

    Article  CAS  Google Scholar 

  65. George, J. et al. Integrative genomic profiling of large-cell neuroendocrine carcinomas reveals distinct subtypes of high-grade neuroendocrine lung tumors. Nat. Commun. 9, 1048 (2018).

    Article  Google Scholar 

  66. Travis, W. D. et al. The 2015 World Health Organization Classification of Lung Tumors: impact of genetic, clinical and radiologic advances since the 2004 classification. J. Thorac Oncol. 10, 1243–1260 (2015).

    Article  Google Scholar 

  67. Guinee, D. G. et al. The spectrum of immunohistochemical staining of small-cell lung carcinoma in specimens from transbronchial and open-lung biopsies. Am. J. Clin. Pathol. 102, 406–414 (1994).

    Article  Google Scholar 

  68. Junker, K., Wiethege, T. & Muller, K. M. Pathology of small-cell lung cancer. J. Cancer Res. Clin. Oncol. 126, 361–368 (2000).

    Article  CAS  Google Scholar 

  69. Kim, Y. H. et al. Combined microarray analysis of small cell lung cancer reveals altered apoptotic balance and distinct expression signatures of MYC family gene amplification. Oncogene 25, 130–138 (2006).

    Article  CAS  Google Scholar 

  70. Barretina, J. et al. The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature 483, 603–607 (2012).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors thank N. Rekhtman for insightful comment regarding pathological criteria for small cell lung cancer diagnosis. This work was supported by grants from the US National Institutes of Health, including U24CA213274 (C.M.R., J.T.P., A.D., J.D.M. and A.F.G.), R01CA197936 (C.M.R., J.T.P. and C.D.), R01CA207295 (L.A.B.), U01CA213273 (L.A.B., J.V.H. and J.S.), P50CA70907 and U01CA213338 (J.E.J., J.D.M. and A.F.G.), U54CA217450 (J.M.L. and V.Q.), UG1CA233259 (J.M.L.) and R21CA216504 (T.G.O.); by Veterans Affairs Merit Review I01CX001425 (P.P.M.); by a LUNGevity Foundation Career Development Award (J.M.L.); and by Cancer Research UK A27412, A25254 and A20465 (C.D.).

Reviewer information

Nature Reviews Cancer thanks T. Bivona, E. Brambilla and other anonymous reviewer(s) for their contribution to the peer review of this work.

Author information

Authors and Affiliations

Authors

Contributions

C.M.R. and J.T.P. researched data for the article, made substantial contributions to the discussion of content, wrote the article and reviewed or edited the article before submission. The other authors all made substantial contributions to the discussion of content and reviewed or edited the article before submission.

Corresponding authors

Correspondence to Charles M. Rudin or John T. Poirier.

Ethics declarations

Competing interests

C.M.R. has consulted for AbbVie, Amgen, Ascentage, AstraZeneca, BMS, Celgene, Daiichi Sankyo, Genentech/Roche, Ipsen, Loxo and PharmaMar; is on the scientific advisory board for Elucida and Harpoon; and receives research funding from Daiichi Sankyo. L.A.B. has consulted for AbbVie, AstraZeneca, BerGenBio, Genmab and PharmaMar and receives research support from AbbVie, AstraZeneca, Genmab and Tolero. C.D. has consulted for AstraZeneca and Merck and receives research funding from AstraZeneca, Epigene, Amgen, FLX Bio, Menarini and Angel. J.V.H. has consulted for AstraZeneca, Boehringer Ingelheim, Exelixis, Genentech, GlaxoSmithKline, Guardant, Hengrui, Lilly, Novartis, Spectrum, EMD Serono and Synta; has received research support from AstraZeneca, Bayer, GlaxoSmithKline and Spectrum; and receives royalties and licensing fees from Spectrum. J.M.L. receives research funding from Ipsen and AbbVie. D.M. received research funding from Janssen and Roche. J.D.M. and A.F.G. receive licensing royalties for cell lines from the US National Institutes of Health and the University of Texas Southwestern Medical Center. J.S. receives research funding from AbbVie. C.R.V. is an adviser to KSQ Therapeutics. All other authors declare no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Rudin, C.M., Poirier, J.T., Byers, L.A. et al. Molecular subtypes of small cell lung cancer: a synthesis of human and mouse model data. Nat Rev Cancer 19, 289–297 (2019). https://doi.org/10.1038/s41568-019-0133-9

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41568-019-0133-9

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer